Research Article GLOBAL MAPPING of ZBTB7A

Total Page:16

File Type:pdf, Size:1020Kb

Research Article GLOBAL MAPPING of ZBTB7A CELLULAR & MOLECULAR BIOLOGY LETTERS http://www.cmbl.org.pl Received: 30 August 2009 Volume 15 (2010) pp 260-271 Final form accepted: 17 February 2010 DOI: 10.2478/s11658-010-0003-7 Published online: 19 March 2010 © 2010 by the University of Wrocław, Poland Research article GLOBAL MAPPING OF ZBTB7A TRANSCRIPTION FACTOR BINDING SITES IN HepG2 CELLS XUYU ZU1, LINGLING YU1, YIMING SUN3, JING TIAN1, FENG LIU1, QINSHENG SUN1, SHENGNAN HE1, GUANG SUN1, WEISHI LUO1 and YUYANG JIANG1,2* 1The Key Laboratory of Chemical Biology, Guangdong Province, Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong 518055, People’s Republic of China, 2School of Medicine, Tsinghua University, Beijing 100084, People’s Republic of China, 3Medical Systems Biology Research Center, Tsinghua University, Beijing 100084, People’s Republic of China, National Engineering Research Center for Beijing Biochip Technology (NERCBBT), 18 Life Science Parkway, Beijing 102206, People's Republic of China Abstract: ZBTB7A is a known proto-oncogene that is implicated in carcinogenesis and cell differentiation and development. Fully understanding the function of ZBTB7A in cellular processes could provide useful strategies for cancer treatment and development-associated disease therapy. Here, global mapping of ZBTB7A transcription factor binding sites was developed by utilizing microarray technology in HepG2 cells. The data obtained from the microarrays was further validated via chromatin immunoprecipitation-PCR (ChIP-PCR) and real time-PCR, and it was revealed that ZBTB7A may be one of the regulators of neural development. ZBTB7A target signal pathways were identified in signal pathway and GO (Gene Ontology) analyses. This is the first report on the global mapping of ZBTB7A downstream direct targets, and these findings will be useful in understanding the roles of ZBTB7A in cellular processes. Key words: ZBTB7A, Proto-oncogene, Transcription factor, ChIP-on-chip analysis, Signal pathway * Author for correspondence. e-mail: [email protected], tel: +86 755 26036017, fax: +86 755 26032094 Abbreviations used: ChIP – chromatin immunoprecipitation; FASN – synthase gene; GO – gene ontology; KEGG – Kyoto Encyclopedia of Genes and Genomes; PMSF – phenylmethylsulfonyl fluoride CELLULAR & MOLECULAR BIOLOGY LETTERS 261 INTRODUCTION ZBTB7A, also known as Pokemon, FBI [1] or LRF [2], is a transcriptional factor that belongs to the POK protein family. The ZBTB7A gene has been shown to play roles in adipogenesis [3], T-lymphoid lineage differentiation, terminal preadipocyte differentiation promotion [2] and carcinogenesis [4, 5]. Homozygous deletion of the ZBTB7A gene resulted in embryonic lethality in a mouse model, indicating that the ZBTB7A gene has an important physiological function. ZBTB7A can self-associate via both the POZ and zinc finger domains [6], and it can also interact with BCL-6, another member of the POK protein family [7]. Considerable effort has been made to identify ZBTB7A target genes in order to elucidate the roles of ZBTB7A in carcinogenesis and cell differentiation and development. It has been shown that FBI-1 represses the transcription of some extracellular matrix genes [8] and suppresses the activity of the ADH5/FDH promoter [9]. Laudes et al. reported that ZBTB7A represses the expression level of E2F4 in a direct mechanism via ZBTB7A regulatory elements within the E2F4 promoter, and that the cyclin A expression level is reduced by ZBTB7A expression through an indirect mechanism without ZBTB7A binding to the promoter of the cyclin A gene [10]. The downregulation of the tumor suppressor Rb gene expression induced by ZBTB7A was found to be associated with the inhibition of C2C12 myoblast cell differentiation [11]. ZBTB7A was proved to be a critical factor in carcinogenesis when it was found to specifically repress the transcription of the tumor suppressor gene ARF through direct binding with the ARF promoter [4]. ZBTB7A was also found to activate the transcription of the fatty acid synthase gene (FASN) together with SREBP-1 [12] . To date, no high-throughput expression profiling had been applied to identify the ZBTB7A target genes. Chip is a powerful tool that identifies direct target genes by isolating DNA bound by proteins. When it was coupled with the microassay detection method (ChIP-chip), direct p53 and c-Myc binding loci on the genomes could be identified [13, 14]. Here, we performed a Zbtb7 ChIP-on-chip study on HepG2 cells, a cell line with a high endogenous ZBTB7A gene expression level, with the aim to delineate the ZBTB7A regulatory network. MATERIALS AND METHODS Cell culture Cells of the human hepatocellular carcinoma cell line HepG2 were purchased from the American Type Culture Collection and were maintained in Dulbecco’s modified Eagle’s medium (DMEM, Gibco BRL) supplemented with 10% heated fetal bovine serum (FBS, Gibco, Grand Island, NY), 2 mM glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin at 37ºC, 5% CO2. Cells at 85-90% confluence were passaged by trypsinization. 262 Vol. 15. No. 2. 2010 CELL. MOL. BIOL. LETT. Chromatin immunoprecipitation and genome-wide ChIP-chip Chromatin immunoprecipitation and genome-wide location analysis were performed as described previously [15]. Briefly, HepG2 cells cultured to a confluence of 80% were treated with formaldehyde (1%) for 30 min, and the cells were collected by centrifugation, washed with ice-cold TBS, and disrupted by vortexing in a lysis buffer. The chromatin was sonicated to yield DNA fragments with an average length of 500 bp. The DNA fragments crosslinked to the proteins were enriched via immunoprecipitation with the ZBTB7A antibody (Abcam, ab36606, Cambridge, MA) and IgG (Abcam, ab37373, Cambridge, MA). After purification, the immunoprecipitated and input DNA were respectively labeled with Cy5 and Cy3 fluorescent dyes via ligation-mediated PCR. Both pools of labeled DNA were hybridized to Affymetrix GeneChip® Tiling Arrays. Images of the Cy5 and Cy3 fluorescence intensities were scanned using a LuxScan-10KA laser confocal scanner (CapitalBio). The signal intensities for each spot were calculated by subtracting the local background using LuxScan 3.0 software (CapitalBio). For each microarray, linear normalization using a per- channel 50th percentile method was adopted to get the ratio for each spot, and any fluorescence ratio over 2-fold higher than the norm was denoted as significant. The data from three independent experiments was combined for a statistical analysis using Student’s t test. Chromatin immunoprecipitation and PCR An independent Chromatin Immunoprecipitation was performed on HepG2 cells, using the same methods as described above. The PCR primers for ZBTB7A target validation are listed in Tab. 1. Tab. 1. PCR primers for ChIP PCR and real time-PCR. Gene Sense (5’-3’) Antisense (5’-3’) Primers for ChIP-PCR CFL1 TTTGCTAGTAGAGTAGGGTG AGAGCCTCAGAAGACACC DPYSL2 TATTCGTGGGAGTTTACC AACAGGTGCTAAGATATGTG LRRC4C TACTTCAGAGAAGTGTTGTAAG GGAATTCAAACAGTCTGC RGS3 GCGAACATGAGCAAGAGG CAAATCCAGAGTCGGGAGAG SEMA4B CAAACCAACGCCACTAGCAG GAGAGCAGGAGCGACGGTGT Primers for real time-PCR CFL1 CTGCCTGAGTGAGGACAAG TTGATGGCGTCCTTGGAG DPYSL2 CCTCGTGTACATGGCTTTC TCCCAGATGACGGACATCC RGS3 CATCTCGGATGAAAGTGCTTTG TCCTTGCATGCCTGGATC SEMA4B GAGGTGAACCGTGAGACAC TGAGGCTGGTAGAGGCTGAC GAPDH AGCCTCAAGATCATCAGCAATG TGTGGTCATGAGTCCTTCCACG Transient transfection of ZBTB7A targeting SiRNA into HePG2 cells Pokemon targeting siRNA (5’-GCUGGACCUUGUAGAUCAAtt-3’, 5’-UUGAU CUACAAGGUCCAGCtt-3’) was synthesized (GeneParma Co, Shanghai, China), and a scramble RNA was used as a control. For the siRNA CELLULAR & MOLECULAR BIOLOGY LETTERS 263 delivery, HepG2 cells were mixed gently with siRNA and OligofectAMINE (Invitrogen, CA) in a volume of 0.5 ml according to the manufacturer’s instructions, and incubated at 37ºC, 5% CO2 for 4 h, followed by the addition of an equal volume of fresh medium containing 20% FBS. The cells were continuously incubated until harvest. Real time-PCR analysis Total RNA was prepared from cells using TRIzol reagent (Invitrogen, CA) according to the manufacturer’s instructions. After being isolated, the RNA was reverse transcribed to cDNA using random primers (Promega, CA). The reaction mixture containing SYBR Green PCR Master Mix (TaKaRa, Dalian, China) was run in a 7500 real-time PCR System (Applied Biosystems, CA). The PCR primers used for quantitative RT-PCR are listed in Tab. 2. The real-time PCR parameters used were as follows: 95ºC for 10 s; 95ºC for 5 s, 60ºC for 34 s for 40 cycles; 95ºC for 15 s, 60ºC for 1 min and 95ºC for 15 s. Tab. 2. Prospective genes identified to be part of the ZBTB7A target signal pathway. Avg. Pathway Gene bank Gene Gene name fold name ID symbol change Aminosugar metabolism NM_003828 myotubularin related protein 1 MTMR1 2.98 NM_000188 hexokinase 1 HK1 2.46 NM_004388 chitobiase CTBS 2.7 NM_003115 UDP-N-acteylglucosamine pyrophosphorylase 1 UAP1 2.03 Epithelial cell signaling in Helicobacter pylori infection nuclear factor of kappa light polypeptide gene enhancer NM_020529 NFKBIA 2.27 in B-cell inhibitor, alpha NM_002751 mitogen-activated protein kinase 11 MAPK11 2.17 NM_006092 caspase recruitment domain family, member 4 CARD4 2.5 protein tyrosine phosphatase, non-receptor type NM_002834 PTPN11 3.06 11 MAPK signaling pathway calcium channel, voltage-dependent, CACNA2 NM_018398 2.6 alpha 2/delta 3 subunit D3 NM_002011 fibroblast growth factor receptor 4 FGFR4 2.14 NM_001394 dual specificity phosphatase 4 DUSP4 3.03 NM_001540 heat shock 27 kDa protein
Recommended publications
  • Two Locus Inheritance of Non-Syndromic Midline Craniosynostosis Via Rare SMAD6 and 4 Common BMP2 Alleles 5 6 Andrew T
    1 2 3 Two locus inheritance of non-syndromic midline craniosynostosis via rare SMAD6 and 4 common BMP2 alleles 5 6 Andrew T. Timberlake1-3, Jungmin Choi1,2, Samir Zaidi1,2, Qiongshi Lu4, Carol Nelson- 7 Williams1,2, Eric D. Brooks3, Kaya Bilguvar1,5, Irina Tikhonova5, Shrikant Mane1,5, Jenny F. 8 Yang3, Rajendra Sawh-Martinez3, Sarah Persing3, Elizabeth G. Zellner3, Erin Loring1,2,5, Carolyn 9 Chuang3, Amy Galm6, Peter W. Hashim3, Derek M. Steinbacher3, Michael L. DiLuna7, Charles 10 C. Duncan7, Kevin A. Pelphrey8, Hongyu Zhao4, John A. Persing3, Richard P. Lifton1,2,5,9 11 12 1Department of Genetics, Yale University School of Medicine, New Haven, CT, USA 13 2Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA 14 3Section of Plastic and Reconstructive Surgery, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA 15 4Department of Biostatistics, Yale University School of Medicine, New Haven, CT, USA 16 5Yale Center for Genome Analysis, New Haven, CT, USA 17 6Craniosynostosis and Positional Plagiocephaly Support, New York, NY, USA 18 7Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA 19 8Child Study Center, Yale University School of Medicine, New Haven, CT, USA 20 9The Rockefeller University, New York, NY, USA 21 22 ABSTRACT 23 Premature fusion of the cranial sutures (craniosynostosis), affecting 1 in 2,000 24 newborns, is treated surgically in infancy to prevent adverse neurologic outcomes. To 25 identify mutations contributing to common non-syndromic midline (sagittal and metopic) 26 craniosynostosis, we performed exome sequencing of 132 parent-offspring trios and 59 27 additional probands.
    [Show full text]
  • Impact of ZBTB7A Hypomethylation and Expression Patterns on Treatment Response to Hydroxyurea Vasiliki Chondrou1†, Eleana F
    Chondrou et al. Human Genomics (2018) 12:45 https://doi.org/10.1186/s40246-018-0177-z PRIMARY RESEARCH Open Access Impact of ZBTB7A hypomethylation and expression patterns on treatment response to hydroxyurea Vasiliki Chondrou1†, Eleana F. Stavrou1†, Georgios Markopoulos2, Alexandra Kouraklis-Symeonidis3, Vasilios Fotopoulos4, Argiris Symeonidis5, Efthymia Vlachaki6, Panagiota Chalkia7, George P. Patrinos8, Adamantia Papachatzopoulou9 and Argyro Sgourou1* Abstract Background: We aimed to clarify the emerging epigenetic landscape in a group of genes classified as “modifier genes” of the β-type globin genes (HBB cluster), known to operate in trans to accomplish the two natural developmental switches in globin expression, from embryonic to fetal during the first trimester of conception and from fetal to adult around the time of birth. The epigenetic alterations were determined in adult sickle cell anemia (SCA) homozygotes and SCA/β-thalassemia compound heterozygotes of Greek origin, who are under hydroxyurea (HU) treatment. Patients were distinguished in HU responders and HU non-responders (those not benefited from the HU) and both, and in vivo and in vitro approaches were implemented. Results: We examined the CpG islands’ DNA methylation profile of BCL11A, KLF1, MYB, MAP3K5, SIN3A, ZBTB7A,andGATA2, along with γ-globin and LRF/ZBTB7A expression levels. In vitro treatment of hematopoietic stem cells (HSCs) with HU induced a significant DNA hypomethylation pattern in ZBTB7A (p*, 0.04) and GATA2 (p*, 0.03) CpGs exclusively in the HU non-responders. Also, this group of patients exhibited significantly elevated baseline methylation patterns in ZBTB7A, before the HU treatment, compared to HU responders (p*, 0.019) and to control group of healthy individuals (p*, 0.021) , which resembles a potential epigenetic barrier for the γ-globin expression.
    [Show full text]
  • Environmental Influences on Endothelial Gene Expression
    ENDOTHELIAL CELL GENE EXPRESSION John Matthew Jeff Herbert Supervisors: Prof. Roy Bicknell and Dr. Victoria Heath PhD thesis University of Birmingham August 2012 University of Birmingham Research Archive e-theses repository This unpublished thesis/dissertation is copyright of the author and/or third parties. The intellectual property rights of the author or third parties in respect of this work are as defined by The Copyright Designs and Patents Act 1988 or as modified by any successor legislation. Any use made of information contained in this thesis/dissertation must be in accordance with that legislation and must be properly acknowledged. Further distribution or reproduction in any format is prohibited without the permission of the copyright holder. ABSTRACT Tumour angiogenesis is a vital process in the pathology of tumour development and metastasis. Targeting markers of tumour endothelium provide a means of targeted destruction of a tumours oxygen and nutrient supply via destruction of tumour vasculature, which in turn ultimately leads to beneficial consequences to patients. Although current anti -angiogenic and vascular targeting strategies help patients, more potently in combination with chemo therapy, there is still a need for more tumour endothelial marker discoveries as current treatments have cardiovascular and other side effects. For the first time, the analyses of in-vivo biotinylation of an embryonic system is performed to obtain putative vascular targets. Also for the first time, deep sequencing is applied to freshly isolated tumour and normal endothelial cells from lung, colon and bladder tissues for the identification of pan-vascular-targets. Integration of the proteomic, deep sequencing, public cDNA libraries and microarrays, delivers 5,892 putative vascular targets to the science community.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage
    The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage Commitment and Effector Function Development in the Immune System This information is current as of September 27, 2021. Aimee M. Beaulieu and Derek B. Sant'Angelo J Immunol 2011; 187:2841-2847; ; doi: 10.4049/jimmunol.1004006 http://www.jimmunol.org/content/187/6/2841 Downloaded from References This article cites 103 articles, 44 of which you can access for free at: http://www.jimmunol.org/content/187/6/2841.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication by guest on September 27, 2021 *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2011 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The BTB-ZF Family of Transcription Factors: Key Regulators of Lineage Commitment and Effector Function Development in the Immune System Aimee M. Beaulieu* and Derek B. Sant’Angelo*,†,‡ Successful immunity depends upon the activity of mul- 1, -2, -4, -5, and -7 (1–11).
    [Show full text]
  • SUPPLEMENTARY NOTE Co-Activation of GR and NFKB
    SUPPLEMENTARY NOTE Co-activation of GR and NFKB alters the repertoire of their binding sites and target genes. Nagesha A.S. Rao1*, Melysia T. McCalman1,*, Panagiotis Moulos2,4, Kees-Jan Francoijs1, 2 2 3 3,5 Aristotelis Chatziioannou , Fragiskos N. Kolisis , Michael N. Alexis , Dimitra J. Mitsiou and 1,5 Hendrik G. Stunnenberg 1Department of Molecular Biology, Radboud University Nijmegen, the Netherlands 2Metabolic Engineering and Bioinformatics Group, Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Athens, Greece 3Molecular Endocrinology Programme, Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Greece 4These authors contributed equally to this work 5 Corresponding authors E-MAIL: [email protected] ; TEL: +31-24-3610524; FAX: +31-24-3610520 E-MAIL: [email protected] ; TEL: +30-210-7273741; FAX: +30-210-7273677 Running title: Global GR and NFKB crosstalk Keywords: GR, p65, genome-wide, binding sites, crosstalk SUPPLEMENTARY FIGURES/FIGURE LEGENDS AND SUPPLEMENTARY TABLES 1 Rao118042_Supplementary Fig. 1 A Primary transcript Mature mRNA TNF/DMSO TNF/DMSO 8 12 r=0.74, p< 0.001 r=0.61, p< 0.001 ) 2 ) 10 2 6 8 4 6 4 2 2 0 Fold change (mRNA) (log Fold change (primRNA) (log 0 −2 −2 −2 0 2 4 −2 0 2 4 Fold change (RNAPII) (log2) Fold change (RNAPII) (log2) B chr5: chrX: 56 _ 104 _ DMSO DMSO 1 _ 1 _ 56 _ 104 _ TA TA 1 _ 1 _ 56 _ 104 _ TNF TNF Cluster 1 1 _ Cluster 2 1 _ 56 _ 104 _ TA+TNF TA+TNF 1 _ 1 _ CCNB1 TSC22D3 chr20: chr17: 25 _ 33 _ DMSO DMSO 1 _ 1 _ 25 _ 33 _ TA TA 1 _ 1 _ 25 _ 33 _ TNF TNF Cluster 3 1 _ Cluster 4 1 _ 25 _ 33 _ TA+TNF TA+TNF 1 _ 1 _ GPCPD1 CCL2 chr6: chr22: 77 _ 35 _ DMSO DMSO 1 _ 77 _ 1 _ 35 _ TA TA 1 _ 1 _ 77 _ 35 _ TNF Cluster 5 Cluster 6 TNF 1 _ 1 _ 77 _ 35 _ TA+TNF TA+TNF 1 _ 1 _ TNFAIP3 DGCR6 2 Supplementary Figure 1.
    [Show full text]
  • Structural Basis of Specific DNA Binding by the Transcription Factor
    8388–8398 Nucleic Acids Research, 2019, Vol. 47, No. 16 Published online 21 June 2019 doi: 10.1093/nar/gkz557 Structural basis of specific DNA binding by the transcription factor ZBTB24 Ren Ren1,†, Swanand Hardikar1,†, John R. Horton1, Yue Lu1, Yang Zeng1,2,AnupK.Singh1, Kevin Lin1, Luis Della Coletta1, Jianjun Shen1,2, Celine Shuet Lin Kong3, Hideharu Hashimoto4, Xing Zhang1, Taiping Chen1,2,* and Xiaodong Cheng 1,2,* 1Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, 2 Houston, TX 77030, USA, Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Downloaded from https://academic.oup.com/nar/article/47/16/8388/5521786 by guest on 23 September 2021 Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA, 3Program in Cancer Biology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA and 4Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA Received October 13, 2018; Revised June 07, 2019; Editorial Decision June 11, 2019; Accepted June 13, 2019 ABSTRACT INTRODUCTION ZBTB24, encoding a protein of the ZBTB family ZBTB (zinc-finger- and BTB domain-containing) proteins, of transcriptional regulators, is one of four known characterized by the presence of an N-terminal BTB genes––the other three being DNMT3B, CDCA7 and (broad-complex, tram-track, and bric-a-brac)` domain [also HELLS––that are mutated in immunodeficiency, cen- known as POZ (poxvirus and zinc finger) domain] and a tromeric instability and facial anomalies (ICF) syn- C-terminal domain with tandem C2H2 Kr¨uppel-type zinc fingers (ZFs), are a large family of evolutionarily con- drome, a genetic disorder characterized by DNA hy- served transcriptional regulators (1).
    [Show full text]
  • Virtual Chip-Seq: Predicting Transcription Factor Binding
    bioRxiv preprint doi: https://doi.org/10.1101/168419; this version posted March 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Virtual ChIP-seq: predicting transcription factor binding 2 by learning from the transcriptome 1,2,3 1,2,3,4,5 3 Mehran Karimzadeh and Michael M. Hoffman 1 4 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada 2 5 Princess Margaret Cancer Centre, Toronto, ON, Canada 3 6 Vector Institute, Toronto, ON, Canada 4 7 Department of Computer Science, University of Toronto, Toronto, ON, Canada 5 8 Lead contact: michael.hoff[email protected] 9 March 8, 2019 10 Abstract 11 Motivation: 12 Identifying transcription factor binding sites is the first step in pinpointing non-coding mutations 13 that disrupt the regulatory function of transcription factors and promote disease. ChIP-seq is 14 the most common method for identifying binding sites, but performing it on patient samples is 15 hampered by the amount of available biological material and the cost of the experiment. Existing 16 methods for computational prediction of regulatory elements primarily predict binding in genomic 17 regions with sequence similarity to known transcription factor sequence preferences. This has limited 18 efficacy since most binding sites do not resemble known transcription factor sequence motifs, and 19 many transcription factors are not even sequence-specific. 20 Results: 21 We developed Virtual ChIP-seq, which predicts binding of individual transcription factors in new 22 cell types using an artificial neural network that integrates ChIP-seq results from other cell types 23 and chromatin accessibility data in the new cell type.
    [Show full text]
  • Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Diseases
    Supplemental Material can be found at: /content/suppl/2018/06/05/70.3.446.DC1.html 1521-0081/70/3/446–474$35.00 https://doi.org/10.1124/pr.117.015354 PHARMACOLOGICAL REVIEWS Pharmacol Rev 70:446–474, July 2018 Copyright © 2018 by The American Society for Pharmacology and Experimental Therapeutics ASSOCIATE EDITOR: ALAN V. SMRCKA Genetic Analysis of Rare Human Variants of Regulators of G Protein Signaling Proteins and Their Role in Human Physiology and Diseases Katherine E. Squires, Carolina Montañez-Miranda, Rushika R. Pandya, Matthew P. Torres, and John R. Hepler Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (K.E.S., C.M.-M., J.R.H.); and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia (R.R.P., M.P.T.) Abstract. ....................................................................................446 I. Introduction. ..............................................................................447 A. G Protein Signaling......................................................................447 B. A (Very) Brief History of Regulators of G Protein Signaling...............................447 C. RGS Protein Rare Human Variants in Complex Diseases . ...............................448 II. RGS Proteins in Physiology and Human Disease . ..........................................449 A. The R4 Family . ........................................................................449 Downloaded from 1. R4 Family Overview..................................................................449
    [Show full text]
  • Supplementary Table 2
    Supplementary Table 2. Differentially Expressed Genes following Sham treatment relative to Untreated Controls Fold Change Accession Name Symbol 3 h 12 h NM_013121 CD28 antigen Cd28 12.82 BG665360 FMS-like tyrosine kinase 1 Flt1 9.63 NM_012701 Adrenergic receptor, beta 1 Adrb1 8.24 0.46 U20796 Nuclear receptor subfamily 1, group D, member 2 Nr1d2 7.22 NM_017116 Calpain 2 Capn2 6.41 BE097282 Guanine nucleotide binding protein, alpha 12 Gna12 6.21 NM_053328 Basic helix-loop-helix domain containing, class B2 Bhlhb2 5.79 NM_053831 Guanylate cyclase 2f Gucy2f 5.71 AW251703 Tumor necrosis factor receptor superfamily, member 12a Tnfrsf12a 5.57 NM_021691 Twist homolog 2 (Drosophila) Twist2 5.42 NM_133550 Fc receptor, IgE, low affinity II, alpha polypeptide Fcer2a 4.93 NM_031120 Signal sequence receptor, gamma Ssr3 4.84 NM_053544 Secreted frizzled-related protein 4 Sfrp4 4.73 NM_053910 Pleckstrin homology, Sec7 and coiled/coil domains 1 Pscd1 4.69 BE113233 Suppressor of cytokine signaling 2 Socs2 4.68 NM_053949 Potassium voltage-gated channel, subfamily H (eag- Kcnh2 4.60 related), member 2 NM_017305 Glutamate cysteine ligase, modifier subunit Gclm 4.59 NM_017309 Protein phospatase 3, regulatory subunit B, alpha Ppp3r1 4.54 isoform,type 1 NM_012765 5-hydroxytryptamine (serotonin) receptor 2C Htr2c 4.46 NM_017218 V-erb-b2 erythroblastic leukemia viral oncogene homolog Erbb3 4.42 3 (avian) AW918369 Zinc finger protein 191 Zfp191 4.38 NM_031034 Guanine nucleotide binding protein, alpha 12 Gna12 4.38 NM_017020 Interleukin 6 receptor Il6r 4.37 AJ002942
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Genomic Anatomy of the Tyrp1 (Brown) Deletion Complex
    Genomic anatomy of the Tyrp1 (brown) deletion complex Ian M. Smyth*, Laurens Wilming†, Angela W. Lee*, Martin S. Taylor*, Phillipe Gautier*, Karen Barlow†, Justine Wallis†, Sancha Martin†, Rebecca Glithero†, Ben Phillimore†, Sarah Pelan†, Rob Andrew†, Karen Holt†, Ruth Taylor†, Stuart McLaren†, John Burton†, Jonathon Bailey†, Sarah Sims†, Jan Squares†, Bob Plumb†, Ann Joy†, Richard Gibson†, James Gilbert†, Elizabeth Hart†, Gavin Laird†, Jane Loveland†, Jonathan Mudge†, Charlie Steward†, David Swarbreck†, Jennifer Harrow†, Philip North‡, Nicholas Leaves‡, John Greystrong‡, Maria Coppola‡, Shilpa Manjunath‡, Mark Campbell‡, Mark Smith‡, Gregory Strachan‡, Calli Tofts‡, Esther Boal‡, Victoria Cobley‡, Giselle Hunter‡, Christopher Kimberley‡, Daniel Thomas‡, Lee Cave-Berry‡, Paul Weston‡, Marc R. M. Botcherby‡, Sharon White*, Ruth Edgar*, Sally H. Cross*, Marjan Irvani¶, Holger Hummerich¶, Eleanor H. Simpson*, Dabney Johnson§, Patricia R. Hunsicker§, Peter F. R. Little¶, Tim Hubbard†, R. Duncan Campbell‡, Jane Rogers†, and Ian J. Jackson*ʈ *Medical Research Council Human Genetics Unit, Edinburgh EH4 2XU, United Kingdom; †Wellcome Trust Sanger Institute, and ‡Medical Research Council Rosalind Franklin Centre for Genome Research, Hinxton CB10 1SA, United Kingdom; §Life Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831; and ¶Department of Biochemistry, Imperial College, London SW7 2AZ, United Kingdom Communicated by Liane B. Russell, Oak Ridge National Laboratory, Oak Ridge, TN, January 9, 2006 (received for review September 15, 2005) Chromosome deletions in the mouse have proven invaluable in the deletions also provided the means to produce physical maps of dissection of gene function. The brown deletion complex com- genetic markers. Studies of this kind have been published for prises >28 independent genome rearrangements, which have several loci, including albino (Tyr), piebald (Ednrb), pink-eyed been used to identify several functional loci on chromosome 4 dilution (p), and the brown deletion complex (2–6).
    [Show full text]